Você está na página 1de 10

OPEN

Citation: Blood Cancer Journal (2016) 6, e441; doi:10.1038/bcj.2016.50


www.nature.com/bcj

REVIEW
‘Acute myeloid leukemia: a comprehensive review and
2016 update’
I De Kouchkovsky1 and M Abdul-Hay1,2

Acute myeloid leukemia (AML) is the most common acute leukemia in adults, with an incidence of over 20 000 cases per year in the
United States alone. Large chromosomal translocations as well as mutations in the genes involved in hematopoietic proliferation
and differentiation result in the accumulation of poorly differentiated myeloid cells. AML is a highly heterogeneous disease;
although cases can be stratified into favorable, intermediate and adverse-risk groups based on their cytogenetic profile, prognosis
within these categories varies widely. The identification of recurrent genetic mutations, such as FLT3-ITD, NMP1 and CEBPA, has
helped refine individual prognosis and guide management. Despite advances in supportive care, the backbone of therapy remains
a combination of cytarabine- and anthracycline-based regimens with allogeneic stem cell transplantation for eligible candidates.
Elderly patients are often unable to tolerate such regimens, and carry a particularly poor prognosis. Here, we review the major
recent advances in the treatment of AML.

Blood Cancer Journal (2016) 6, e441; doi:10.1038/bcj.2016.50; published online 1 July 2016

INTRODUCTION absence of any large chromosomal abnormality.7 Studies of animal


Acute myeloid leukemia (AML) is the most common acute leukemia models at the turn of the century led to the development of
in adults, accounting for ~ 80 percent of cases in this group.1 Within a two-hit model of leukemogenesis, which offers a conceptual
the United States, the incidence of AML ranges from three to five framework for classifying the various mutations associated with
cases per 100 000 population. In 2015 alone, an estimated 20 830 AML.8 According to this model, class I mutations which result in the
new cases were diagnosed, and over 10 000 patients died from this activation of pro-proliferative pathways must occur in conjunction
disease.2 The incidence of AML increases with age, from ~ 1.3 per with class II mutations which impair normal hematopoietic
100 000 population in patients less than 65 years old, to 12.2 cases differentiation in order for leukemia to develop.9,10 Common class
per 100 000 population in those over 65 years. Although advances I mutations, such as FLT3 (internal tandem duplications, ITD, and
in the treatment of AML have led to significant improvements in tyrosine kinase domain mutations, TKD), K/NRAS, TP53 and c-KIT are
outcomes for younger patients, prognosis in the elderly who found in ~ 28, 12, 8 and 4% of cases, respectively.7 Studies of solid
account for the majority of new cases remains poor.3 Even with and hematological malignancies have also highlighted the role
current treatments, as much as 70% of patients 65 years or older will of signal transducer and activator of transcription 3 (STAT3) in the
die of their disease within 1 year of diagnosis.4 stimulation of cellular proliferation and survival.11–13 Enhanced
tyrosine phosphorylation of STAT3 whether due to increased
secretion of cytokines, such as IL-6(ref. 14) or mutations in receptor
PATHOPHYSIOLOGY tyrosine kinases (for example, FLT3 duplications15 or less frequently
AML can arise in patients with an underlying hematological JAK2)16 is seen in up to 50% of AML cases and signifies a worse
disorder, or as a consequence of prior therapy (for example, prognosis. Notable class II mutations include NPM1 and CEBPA,
exposure to topoisomerases II, alkylating agents or radiation).5 which are found in ~ 27% and 6% of cases, respectively, and confer
However in majority of cases, it appears as a de novo malignancy a better prognosis.7 Alterations in genes involved in epigenetic
in previously healthy individuals. Regardless of its etiology, regulation have recently emerged as a third class of mutations, with
the pathogenesis of AML involves the abnormal proliferation downstream effects on both cellular differentiation and prolifera-
and differentiation of a clonal population of myeloid stem cells. tion. These include mutations in the DNA-methylation related genes
Well-characterized chromosomal translocations, such as t(8:21) in DNMT3A, TET2, and IDH-1 and IDH-2,6,7 which are found in more
core-binding factor AML (CBF-AML) or t(15:17) in acute promye- than 40% of AML cases.
locytic leukemia (APL) result in the formation of chimeric proteins Despite significant advances, much remains to be discovered
(RUNX1-RUNX1T1 and PML-RARA, respectively), which alter the on the exact contribution of these individual mutations
normal maturation process of myeloid precursor cells. In addition to the development of AML. As suggested by the ‘two-hit model,’
to large chromosomal rearrangements, molecular changes have the pathogenesis and behavior of AML depends heavily on
also been implicated in the development of AML. In fact, genetic the interactions between different somatic alterations and
mutations are identified in more than 97% of cases,6 often in the chromosomal rearrangements. Thus, the c-KIT mutation has been

1
Department of Medicine, New York University School of Medicine, New York, NY, USA and 2Department of Hematology/Oncology, New York University Perlmutter Cancer
Center, New York, NY, USA. Correspondence: Dr M Abdul-Hay, Department of Hematology/Oncology, NYU School of Medicine, New York Perimutter Cancer Center, 240 East 38th
Street, 19 Floor, New York, NY 10016, USA.
E-mail: Maher.Abdulhay@nyumc.org
Received 13 April 2016; revised 3 May 2016; accepted 19 May 2016
AML: a comprehensive review and 2016 update
I De Kouchkovsky and M Abdul-Hay
2
associated with t(8;21) or inv(16), and its presence carries significant morphology, immunophenotype and clinical presentation to
implications regarding prognosis. Similarly, NMP1 (a class II mutation) define six major disease entities: AML with recurrent genetic
frequently occurs in conjunction with the class I mutation FLT3-ITD, abnormalities; AML with myelodysplasia-related features; therapy-
or mutations in the epigenetic genes DNMT3A and IDH-1 or IDH-2.6 related AML; AML not otherwise specified; myeloid sarcoma; and
Most of the clinical manifestations of AML reflect the accumulation myeloid proliferation related to Down syndrome (Table 1).19
of malignant, poorly differentiated myeloid cells within the bone Among cases of AML with recurrent genetic abnormalities, 11
marrow, peripheral blood and infrequently in other organs. The subtypes are further delineated according to distinct chromoso-
majority of patients presents with a combination of leuko- mal translocations. In addition, the provisional entities AML with
cytosis and signs of bone marrow failure such as anemia and mutated NPM1 and AML with mutated CEBPA were introduced
thrombocytopenia. Fatigue, anorexia and weight loss are common as part of the 2008 revision,18 while AML with BCR-ABL1 and
complaints; lymphadenopathy and organomegaly are not typically AML with mutated RUNX1 were introduced as part of the 2016
seen. If left untreated, death usually ensues within months revision.19 Genetic abnormalities also inform the diagnosis of AML
of diagnosis secondary to infection or bleeding. The diagnosis with myelodysplasia-related changes: along with a history of MDS
of acute leukemia is established by the presence of 20% or more or morphological evidence of dysplasia in two or more myeloid
blasts in the bone marrow or peripheral blood.17 AML is further cell lineages, the presence of myelodysplasia-related cytogenetic
diagnosed by demonstrating the myeloid origin of these cells abnormalities such as monosomy 5 or 7, and deletion 5q or 7q
through testing for myeloperoxidase activity, immunophenotyp- identify cases of AML with myelodysplasia-related features.
ing or documenting the presence of Auer rods. The latter finding
consists of azurophilic, often needle-shaped cytoplasmic inclusion
bodies that are commonly seen in APL, acute myelomonocytic PROGNOSTIC FACTORS
leukemia and the majority of AML with t(8;21). The diagnosis of Accurate assessment of prognosis is central to the management
AML can also be established in the presence of an extramedullary of AML. By stratifying patients according to their risk of treatment
tissue infiltrate, or a documented t(8;21), inv(16) or t(15;17) in the resistance or treatment-related mortality (TRM), prognostic factors
appropriate clinical setting, regardless of the blast percentage.18 help guide the physician in deciding between standard or
increased treatment intensity, consolidation chemotherapy or
allogenic hematopoietic stem cell transplant, or more fundamen-
CLASSIFICATION tally in choosing between established or investigational therapies.
The French–American–British classification system represents Among clinical factors, increased age and poor performance
the first attempt to distinguish between different types of AML. status are both associated with lower rates of complete remission
Established in 1976, it defines eight subtypes (M0 through M7) (CR) and decreased overall survival (OS).3,20 Age and performance
based on the morphological and cyto-chemical characteristics status at diagnosis similarly help to predict the risk of TRM,
of the leukemic cells. In 2001, as part of an effort to integrate although multivariate model analyses suggest that other variables
advances made in the diagnosis and management of AML, the such as platelet count, serum creatinine or albumin rather than
World Health Organization (WHO) introduced a new classification age itself account for most of the increased risk of TRM seen
system followed by a revised version in 2008.18 Later in 2016 in older patients.21 Therapy-related AML and AML associated with
a new revised version was released, the WHO classification of AML a prior hematological malignancy also carry a significantly poorer
distinguishes itself by incorporating genetic information with prognosis.22 Although clinical factors have an important role in

Table 1. WHO classification of AML and related neoplasms

Types Genetic abnormalites

AML with recurrent genetic abnormalities AML with t(8:21)(q22;q22); RUNX1-RUNX1T1


AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11
APL with PML-RARA
AML with t(9;11)(p21.3;q23.3); MLLT3-KMT2A
ML with t(6;9)(p23;q34.1); DEK-NUP214
AML with inv(3)(q21.3q26.2) or t(3;3)(q21.3;q26.2); GATA2, MECOM
AML (megakaryoblastic) with t(1;22)(p13.3;q13.3); RBM15-MKL1
AML with BCR-ABL1 (provisional entity)
AML with mutated NPM1
AML with biallelic mutations of CEBPA
AML with mutated RUNX1 (provisional entity)
AML with myelodysplasia-related changes
Therapy-related myeloid neoplasms
AML with minimal differentiation
AML without maturation
AML with maturation
Acute myelomonocytic leukemia
Acute monoblastic/monocytic leukemia
Acute erythroid leukemia
Pure erythroid leukemia
Acute megakaryoblastic leukemia
Acute basophilic leukemia
Acute panmyelosis with myelofibrosis
Myeloid sarcoma
Myeloid proliferations related to Down syndrome Transient abnormal myelopoiesis
ML associated with Down syndrome
Abbreviations: AML, acute myeloid leukemia; APL, acute promyelocytic leukemia; ML, myeloid leukemia; WHO, World Health Organization.

Blood Cancer Journal


AML: a comprehensive review and 2016 update
I De Kouchkovsky and M Abdul-Hay
3
guiding therapy, cytogenetic changes constitute the single stron- cytogenetic and a complex karyotype.36 However regardless
gest prognostic factor for CR and OS in AML. Accordingly, cases of the cytogenetic profile, TP53 mutations are associated with
of AML can be stratified into favorable, intermediate or adverse a very poor prognosis10 and may in fact represent the single worst
prognostic risk groups based on their cytogenetic profile alone. genetic prognostic factor.36 Mutations in DNA-related genes
The chromosomal rearrangements t(8;21), t(15;17) or inv(16) all also carry important implications for the prognosis and treatment
confer a favorable prognosis,17,23 with a 3 year OS of 66% and 33% of AML. The presence of a mutated DNA methyl transferase
in patients younger and older than 60 years, respectively.24 In gene DNMT3A has been associated with a worsened prognosis
contrast, cytogenetic changes such as a complex karyotype (that is, in CN-AML and adverse-risk AML.37 Partial tandem duplica-
three or more chromosomal abnormalities in the absence of tions of KMT2A (previously known as MLL), which encodes a
any of the recurrent genetic abnormalities identified in the WHO histone methyltransferase, have also been associated with a worse
2008 classification), monosomy 5 or 7, t(6;9), inv(3) or 11q changes prognosis in CN-AML.6,10,36 The prognostic impact of IDH-1/IDH-2
other than t(9;11) have all been associated with a significantly mutations is less well established and is likely modified by
higher risk of treatment failure and death (Table 2).24 AML cases co-occurring mutations. Among cases of FLT3-ITD-negative and
with an intermediate prognostic risk mainly constitute of patients NPM1-mutated CN-AML, IDH-1/IDH-2 mutations have been
with normal cytogenetics (CN-AML).17,24 shown to improve OS.6 However a recent study of 826 patients
Gene mutations have helped further refine risk stratification with known IDH-1 and IDH-2 status found no prognostic impact
based on cytogenetic changes alone. Among patients with t(8;21), on treatment response or OS.38 Further analysis is required
the presence of a c-KIT mutation significantly increases the risk to delineate the role of DNA-related genes in OS and treatment
of relapse, and decreases OS to levels comparable to those of response. In addition to genetic profiling at the time of diagnosis,
patient with intermediate-risk AML.6,23,25 Although there is some information gained after treatment initiation plays a growing
evidence that the presence of c-KIT mutations similarly lowers role in refining patient prognosis: As could be expected,
prognosis in patients with inv(16),26 recent studies have failed individuals who achieve CR (defined morphologically as a blast
to show any prognostic impact in this subset of cases.6,27,28 count of o5% of total nonerythroid cells in the bone marrow) after
Molecular changes have a particularly important role in refining induction therapy have a significantly increased survival compared
the prognosis of patients with CN-AML, which includes nearly with patients with treatment resistant AML.39,40 Survival among
half of de novo AML cases. Thus CN-AML with a mutated CEBPA patients achieving CR is further influenced by the correction or
or a mutated NPM1 in the absence of FLT3-ITD has been identified persistence of thrombocytopenia, with a shorter duration of survival
as having a prognostic risk similar to that of AML with favorable observed in the latter group.40 More recently, techniques such
cytogenetic changes.17,29 The favorable prognostic impact as real-time PCR and flow cytometry have been used to measure
of CEBPA mutations has been further refined to biallelic mutations the presence of minimal residual disease among patients in CR.
only.30 On the other hand, multiple studies29,31 including a Persistently elevated levels of RUNX1-RUNX1T1 transcripts after
meta-analysis of relapse-free survival (RFS) and OS in patients with induction therapy in patients with t(8;21) AML are thus associated
CN-AML o 60 years of age have consistently shown the presence with an increased incidence of relapse.41,42 Similarly among patients
of FLT3-ITD to be associated with a worsened prognosis.6,32 This with intermediate-risk disease, detection of minimal residual disease
has led to the classification of CN-AML with FLT3-ITD into by flow cytometry is an independent predictor of relapse and
the adverse prognostic-risk group.23,33 As with CEBPA mutations, survival43,44 and carries important implications for management.45
the prognostic impact of FLT3-ITD may depend on the presence
of biallelic mutations. Several studies have shown a significantly
worse prognosis in patients with higher mutant to wild-type ESTABLISHED TREATMENTS
allelic ratios.34,35 TP53 mutations, which are found in only 2– 8% Eligible patients first undergo induction therapy to achieve CR.
of cases,6,7 occur more frequently in cases with unfavorable Unfortunately, minimal residual disease often persists in CR, and

Table 2. Prognostic-risk group based on cytogenetic and molecular profile

Prognostic-risk group Cytogenetic profile alone Cytogenetic profile and molecular abnormalities

Favorable t(8:21)(q22;q22) t(8:21)(q22;q22) with no c-KIT mutation


inv(16)(p13;1q22) inv(16)(p13;1q22)
t(15;17)(q22;q12) t(15;17)(q22;q12)
Mutated NPM1 without FLT3-ITD (CN-AML)
Mutated biallelic CEBPA (CN-AML)
Intermediate CN-AML t(8:21)(q22;q22) with mutated c-KIT
t(9;11)(p22;q23) CN-AML other than those included in the
favorable or adverse prognostic group
Cytogenetic abnormalities not included in the t(9;11)(p22;q23)
favorable or adverse prognostic risk groups
Cytogenetic abnormalities not included in the favorable
or adverse prognostic risk groups
Adverse inv(3)(q21q26.2) TP53 mutation, regardless of cytogenetic profile
t(6;9)(p23;q34) CN with FLT3-ITD
11q abnormalities other than t(9;11) CN with DNMT3A
− 5 or del(5q) CN with KMT2A-PTD
−7 inv(3)(q21q26.2)
Complex karyotype t(6;9)(p23;q34)
11q abnormalities other than t(9;11)
− 5 or del(5q)
−7
Complex karyotype
Abbreviations: AML, acute myeloid leukemia; ITD, internal tandem duplications.

Blood Cancer Journal


AML: a comprehensive review and 2016 update
I De Kouchkovsky and M Abdul-Hay
4
relapse will inevitably occur if treatment is discontinued. There- and prevent relapse. Available options for consolidation include
fore, a favorable response to induction therapy should be followed chemotherapy and allogeneic hematopoietic stem cell transplant
by consolidation therapy in order to eradicate any residual (allo-HSCT). When choosing between these different options, the
disease and achieve lasting remission. The mainstay of induction risk of TRM should be weighted against the risk of treatment
therapy consists of the ‘7+3’ regimen, which combines 7 days failure or relapse. Intention-to-treat analyses (allocating patients
of continuous infusion cytarabine with 3 days of anthracycline. to allo-HSCT or chemotherapy based on the availability of a related-
It is generally offered to patients with an intermediate to favorable donor) have found no benefit to allo-HSCT when compared with
prognosis and a low risk of TRM (for example, younger patients chemotherapy in patients with cytogenetically favorable AML in first
with good performance status, normal creatinine, albumin and CR.61,62 Thus chemotherapy is a reasonable first-line consolidation
platelet count).23 Studies of induction regimens using either choice for patients with a favorable prognosis. Regimens generally
daunorubicin at 60 or 90 mg/m2, or idarubicin at 12 mg/m2 consist of intermediate-dose cytarabine (two to four cycles each
have shown similar rates of CR and survival.23,46,47 A subset of consisting of six doses at 1.5–3 g/m2), which has been shown to be as
patients with DNMT3A and KMT2A mutations, which represents effective as high-dose cytarabine48,50,63 or multi-agent regimens.50
a poor prognostic marker, may however benefit from higher The optimal choice of consolidation therapy for patients with
doses of daunorubicin.6 Standard dosing of cytarabine consists an intermediate-risk cytogenetic profile but favorable genetic
of 100–200 mg/m2 daily administered as a continuous infusion mutations is more controversial: several studies have found
over 7 days. Although studies have shown greater efficacy at no benefit to transplantation in patients with NPM1-mutated,
higher doses, this added benefit is small and accrued at the cost FLT3-ITD-negative CN-AML.64,65 However in a recent intention-to-
of increased toxicity;23,48,49 induction therapy with high-dose treat analysis, allo-HSCT was shown to improve RFS in this
cytarabine is generally reserved for refractory disease. The subset of patients.66 While these conflicting results may reflect
combination of fludarabine, cytarabine, G-CSF and idarubicin differences in study design, the difference in observed outcomes
(FLAG-IDA), which was traditionally used for the treatment may also reflect the modulating effect of co-occurring mutations,
of relapse, has also been shown to be a reasonable alternative such as IDH-1/-2.6 On the other hand allo-HSCT significantly
to standard induction regimens and results in similar CR rates and prolongs RFS and OS in some patients with intermediate-risk and
OS overall but higher rates of CR after a single course.50 in most with adverse-risk AML, and should be offered as a first-line
An optimal approach to elderly patients with AML has not consolidation therapy in eligible patients.62,67–69 In addition, allo-
been established. Individuals over the age of 65 are more likely HSCT has been shown to prolong RFS and improve OS in patients
to present with an adverse cytogenetic-risk profile, are less likely with CN-AML and a high FLT3-ITD allelic ratio.70
to respond to chemotherapy and are often more susceptible to
treatment-related toxicities. However despite a significantly worse
prognosis, induction therapy improves survival in patients over NOVEL AGENTS
the age of 65 when compared with supportive care and palliative FLT3-ITD inhibitors
chemotherapy,51 and should be pursued whenever possible.
Hypomethylating agents, traditionally used for the treatment Inhibition of tyrosine kinase (TK) receptors has been used
of myelodysplastic syndrome (MDS), have also shown efficacy successfully in various solid and hematological malignancies,
in elderly patients with AML. Evidence of a therapeutic benefit was including Philadelphia-chromosome positive leukemias. Given the
first demonstrated in post hoc analyses of patients with MDS who prognostic impact and the high rate of FLT3 mutations, inhibition
were retrospectively found to meet diagnostic criteria for AML of this TK has long been recognized as a potential therapeutic
under the WHO classification.52 A 2012 randomized trial of the target in AML. Tested agents include the first-generation inhibitors
hypomethylating agent decitabine versus supportive care or low- sorafenib and midostaurin, as well as newer second-generation
dose cytarabine in patients 65 years or older showed a significant agents such as quizartinib and crenolanib.
improvement in OS with hypomethylating therapy (although
this survival advantage failed to meet statistical significance in Sorafenib. Sorafenib is a tyrosine kinase inhibitors (TKI) of RAF
the primary analysis).53 A more recent trial comparing the kinase, c-KIT, VGFR, PGFR and FLT3-ITD, which was first used
hypomethylating agent azacitidine to supportive care, low-dose for the treatment of hepatocellular and renal cell carcinoma. As early
cytarabine or standard induction therapy in patients 65 years or as 2008, phase I trials of sorafenib administered as a single agent
older did not show any significant improvement in median OS.54 n patients with FLT3-ITD-positive relapsed or refractory (r/r) AML
However among patients pre-selected to receive supportive demonstrated significant reductions in the number of leukemic cells
care, a subgroup analysis suggested a benefit to azacitidine both in the peripheral blood and bone marrow, achieving CR in
therapy. A similar benefit was seen among patients with adverse several patients.71–74 In a phase II trial of 13 patients with r/r FLT3-
cytogenetic-risk profile or MDR-AML.54 These results suggest ITD-positive AML, single-agent sorafenib at doses of 200–400 mg
a promising role for the use of hypomethylating agents in older twice daily established CR (including CR with insufficient hemato-
individuals, including as a bridge for induction chemotherapy with logic recovery) in over 90% of cases. The agent was well-tolerated,
the goal of achieving CR. with grades 3 to 4 adverse events consisting of hyperbilirubinemia
Response to induction therapy should be evaluated 14 days (in 4/13 patients), elevated transaminases (5/13), diarrhea (4/13),
after initiation of treatment with a bone marrow aspirate and core rash (2/13), pancreatitis (1/13), colitis (1/13), pericarditis (1/13),
biopsy.17 Up to 25–50% of patients show persistent cytological hand and foot syndrome (2/13) and elevated creatinine (1/13).
evidence of disease after one cycle of standard induction therapy Yet despite a strong initial response, the majority of patients
and require reinduction.55 Treatment options for such patients relapsed within 72 days of remission. Treatment failure was
(as well as patients with a disease relapse) include a second cycle associated with the emergence of D835Y and D835H mutations
of standard dose cytarabine combined with an anthracycline, within the FLT3 TKD. The addition of sorafenib to standard induction
high-dose cytarabine alone or FLAG-IDA, with roughly similar CR regimens has yielded similarly mixed results: although initial phases I
rates of up to 50%.23,56,57 In addition, mitoxantrone-based regimens and II trials of combination therapy were able to achieve longer
(in combination with etoposide and/or cytarabine) have been shown periods of disease-free survival, relapse invariably ensued within
to achieve CR up to 40–60% of patients with recurrent or refractory several months of treatment.75–77 In one such study, the combina-
AML.58,59 Ultimately, around 60–80% of patients with de novo AML tion of sorafenib with cytarabine and idarubicin as induction and
will achieve CR with induction therapy.60 Patients in remission consolidation therapy was able to achieve CR (or CR with an
should be offered consolidation therapy to eradicate residual disease incomplete platelet recovery) in 18 out of 18 patients with previously

Blood Cancer Journal


AML: a comprehensive review and 2016 update
I De Kouchkovsky and M Abdul-Hay
5
untreated FLT3-ITD-positive AML.75 However after a median follow-up Midostaurin. Midostaurin is another first-generation FLT3 TKI
of 9 months, more than half of these patients had relapsed. with significant but transient single-agent activity in patients
Interestingly, no new mutation was observed in the FLT3 TKD of with AML.86,87 As with sorafenib, its effects are limited by the
the relapsed samples available for genetic sequencing. Alternative rapid emergence of resistance. Combinations of midostaurin
mechanisms of resistance, such as the increased levels of FLT3 and existing chemotherapy regimens are currently under inves-
receptor ligand seen in patients receiving standard chemotherapy, tigation: results of a phase I and combined phase I/II trial of
have been postulated to contribute to treatment failure.78 Combina- midostaurin and azacitidine have recently been published,
tions of sorafenib and hypomethylating agents, which have not been demonstrating the tolerability and efficacy of this combination in
associated with an increase in FLT3 receptor ligand levels, are currently patients with AML. In their cohort of 17 patients with a median age
being investigated. An encouraging phase II trial of sorafenib and of 73, Cooper et al.88 established a MTD level of 75 mg PO twice
azacitidine in 43 patients with relapsed/refractory AML reported a daily with no observed dose-limiting toxicities. In another phase I/II
response rate of 46%, including 16% CR and 27% CR with incomplete trial of the combination of midostaurin and azacitidine was tested
count recovery.79 in patients with AML (primary or secondary) and MDS, Strati et al.
The role of sorafenib as a first-line therapy in AML was further reported a lower MTD of 50 mg PO twice daily. The combination
delineated in two recent randomized trials: In 2013, Serve et al. achieved an overall response rate of 26%, with median remission
published the results of study, in which 201 patients 60 years or duration of 20 weeks. Although no DLT were observed in this
older with newly diagnosed AML were randomized to receive study, neutropenia, thrombocytopenia and anemia developed
sorafenib or placebo in addition to standard chemotherapy. in 96, 94 and 61% of patients, as well as infections and a decreased
Even within the subset of patients with FLT3-ITD-positive disease, left ventricular ejection fraction in 56 and 11% of patients,
the addition of sorafenib did not result in an improved EFS or OS, respectively.89 In a recent randomized, double-blind trial of 717
and was instead associated with an increased incidence of adverse patients with previously untreated FLT3 (ITD and TKD) positive AML,
events.80 Most recently, Rollig et al.81 investigated the combina- Stone et al. explored the role of midostaurin in combination with
tion of sorafenib with standard chemotherapy in a multicenter a standard ‘7+3’ induction regimen and high-dose cytarabine
randomized controlled phase II trial of 267 patients age 60 consolidation therapy. Patients randomized to the midostaurin
or younger with newly diagnosed AML. In this study, patients treatment arm also received midostaurin as maintenance therapy
were randomly assigned to receive two cycles of standard ‘7+3’ for one year. Although no difference in the rate of CR was observed
induction therapy followed by three cycles of high-dose cytarabine between the midostaurin and placebo arms, patients receiving
as consolidation therapy in combination with either sorafenib midostaurin had a significantly higher OS and EFS (with a hazard
(400 mg twice daily) or placebo. Patients assigned to the sorafenib ratio of 0.77 and 0.80, respectively).90
group also received 12 months of sorafenib maintenance therapy
after their last consolidation cycle. After 3 years the primary end Quizartinib. Second-generation inhibitors such as quizartinib
have been designed to specifically target the FLT3 kinase, in order
point, event-free survival, was achieved in 40% of patients in the
to reduce toxicity from off-target effects. In addition to this
sorafenib group, versus 20% in the placebo group (with an
increased selectivity, quizartinib also possesses a good bioavail-
unadjusted hazard ratio of 0.64).81 The occurrence of grades 3–5
ability and a half-life of more than 24 h, which allows for a more
diarrhea, rash, fever and bleeding were significantly increased
continuous FLT3 inhibition. A phase I study of oral quizartinib in
among patients receiving sorafenib (occurring in 11, 7, 54 and 7%,
76 patients with relapsed/refractory AML was able to achieve
respectively). Importantly, only 17% of the individuals enrolled a hematological response in 30% of patients, and a CR in 13%
in this study were positive for the FLT3-ITD mutation. The observed regardless of FLT3 mutational status. Among patients with FLT3-ITD,
benefit in FLT3-ITD-negative AML may in part be explained by off- the rate of hematologic response increased to 53%, with ~ 23%
target inhibition of other tyrosine kinases, such as c-KIT, PGFR and of patients achieving CR. Patients were able to tolerate doses of
RAF kinase. Alternatively as suggested by the observed increased up to 200 mg/day, with grade 3 QT interval prolongation as the
in FLT3 receptor ligands level activation of wild-type FLT3 TK may only DLT.91 In one phase 2 trial, 137 patients with relapsed/
become a driver of leukemogenesis in patients receiving standard refractory AML were given quizartinib monotherapy. The agent was
chemotherapy.82 By targeting FLT3 TK in the period immediately administered in 28-day cycles at doses of 90 mg/day in females
after cytarabine or daunorubicin therapy, sorafenib may exert and 135 mg/day in males; the most common treatment toxicities
significant anti-leukemic activity even in FLT3-ITD-negative cells. were nausea and vomiting in 38 and 26% of patients, anemia in
In an interesting parallel to the use of TKI in Philadelphia- 29%, QT interval prolongation in 26%, febrile neutropenia in 25%,
chromosome-positive leukemias, inhibition of FLT3 TK has also diarrhea in 20% and fatigue in 20%. The rate of composite CR
shown a promising role in the post-allo-HSCT setting, either as (that is, CR, CR with incomplete platelet recovery and CR with
maintenance therapy or treatment of relapse. This was explored incomplete hematological recovery) and the median OS were 34%
recently by Chen et al.83 in a phase I trial of 22 patients with and 25.6 months in FLT3-ITD-negative cases, and 44% and
FLT3-ITD-positive disease who received sorafenib as maintenance 23.1 months in FLT3-ITD positive cases. The observed benefit in
therapy following allo-HSCT. In addition to establishing safety FLT3-ITD-negative disease may be explained by off-target effects,
and feasibility, the authors of this study reported rates of PFS and or by the upregulation of the FLT3 TK pathway as suggested by the
OS which compared favorably to historical controls, particularly rise in FLT3 receptor ligand levels in patients receiving standard
in the subset of patients in CR1 or CR2 at the time of transplant. chemotherapy.92 Once again however, the response to FLT3 TKI is
In a retrospective analysis of six patients status post allo-HSCT, limited by the rapid emergence of resistance: the median duration
sorafenib as maintenance therapy (n = 5) or treatment of relapse of remission was only 5 weeks in patients with FLT3-ITD-positive
(n = 1) resulted in a median PFS of 16 months, with all patients AML.93 Interim analysis of a phase I/II trial of quizartinib in
remaining in molecular remission (that is, FLT3-ITD-negative combination with azacitidine or cytarabine (NCT01892371) reported
by PCR).84 Interestingly, skin graft-versus-host-disease occurred an overall response rate of 82% in FLT3-ITD-positive AML, MDS or
shortly after initiation of therapy in five out of these six patients. chronic myelomonocytic leukemia.94 A phase III trial comparing
In addition to its action as a TKI, sorafenib may therefore possess quizartinib monotherapy to salvage chemotherapy in relapsed and
an immunomodulatory role, which synergizes with the graft- refractory AML (NCT02039726) is similarly underway.
versus-leukemia effect.85 Two phase I trials of sorafenib use in the
peri-transplant setting are currently underway (NCT01398501 and Crenolanib. Crenolanib besylate is an orally available second-
NCT01578109). generation FLT3 TKI, with activity against FLT3-ITD and FLT3-TKD

Blood Cancer Journal


AML: a comprehensive review and 2016 update
I De Kouchkovsky and M Abdul-Hay
6
mutants. Unlike other FLT3 TKIs, which are subject to the it was reported a CR rate of 63% using the combination of
emergence of resistance conferring kinase domain mutations clofarabine with low-dose cytarabine, compared with 31% with
(such as D835Y), crenolanib appears to possess extensive ‘pan- clofarabine alone.107 In a study of 320 patients over the age of 55
kinase’ inhibition of secondary TKD mutations. Using concentra- with relapsed/refractory AML, the combination of clofarabine and
tions far below the clinically achievable plasma levels, Smith et al. cytarabine achieved significantly higher rates of CR, CR with
were unable to identify any single TKD mutation able to confer incomplete platelet count and DFS when compared with cytarabine
resistance to crenolanib.95 In a phase II study of 38 patients with alone.108 Although neither of these studies was able to show
FLT3-mutated AML (including relapsed and refractory patients), an improved OS, these results suggest a synergistic action between
crenolanib administered at doses of 200 mg/m2 per day three clofarabine and cytarabine, and have spurred interest in the
times a day in 28 days cycle achieved a median EFS and OS of combination of these two agents. Recently a study was published
8 and 19 weeks, respectively.96 Crenolanib is currently being showing the results of a phase 2 trial of clofarabine and low-dose
studied in multiple clinical trials in AML patients, both with and cytarabine in older patients with newly diagnosed AML.109 In this
without FLT3-mutated AMLs.
study, 118 patients age 60 or older (median age of 68 years)
received induction therapy with clofarabine at 20 mg/m2 on day 1
STAT inhibitors through 5 and low-dose cytarabine at 20 mg subcutaneously
STAT3 tyrosine phosphorylation is upregulated in up to 50% of AML twice daily on day 1 through 10. In an attempt to improve survival,
cases and confers a worse prognosis. Activation of the STAT3 this was followed by consolidation and maintenance therapy with
signaling pathway is also stimulated by the FLT3 receptor ligand,97 up to 18 cycles of clofarabine and low-dose cytarabine alternating
and may represent a key step in the development of FLT3 TKI with decitabine. CR was achieved in 60% of cases, with a median
resistance. Several small molecules of STAT3 inhibitors have been OS of 11.1 months, and a median RFS of 14.1 months. The regimen
developed and are currently being investigated for the treatment was well-tolerated, with a 4-week mortality rate of 3%. The most
of AML: in 2011, Redell et al. showed decreased STAT3 phosphory- common non-hematological toxicities reported in this study
lation and induction of apoptosis in AML cell lines treated with the included nausea in 81% of cases, elevated liver transaminases
STAT3 inhibitor C188-9.98 More recently, the optimized compound and bilirubin in 64 and 47% of cases, as well as rash in 56% of
MM-206 demonstrated a dose-dependent induction of apoptosis
cases. Clofarabine may thus represent a well-tolerated addition to
in AML cell lines cultured in the presence of bone marrow stromal
low-dose cytarabine in elderly patients unable to receive standard
cells. The anti-tumor activity of MM-206 was confirmed in vivo
chemotherapy or allo-HSCT for consolidation therapy. Clofarabine
by reducing blast count and improving survival in AML-engrafted
mice.99 OPB-31121 is a small molecule inhibitor of STAT3 and STAT5 has shown similarly promising results in younger patients when
phosphorylation, which has demonstrated activity in advanced combined with the standard ‘7+3’ induction regimen. In a study of
solid tumors.100 Treatment of various leukemic cell lines with this 57 newly diagnosed patients under the age of 60, the combination
compound resulted in significant growth inhibition, including of clofarabine (at 22.5 mg/m2 IV daily for 5 days) with idarubicin
FLT3-ITD-positive AML cells.101 Importantly, OPB-31121 was able and cytarabine used as a frontline induction and consolidation
to overcome FLT3 receptor ligand-induced STAT3 phosphoryla- therapy achieved CR rates of 74% and a median EFS of 13.5 months
tion, and may help to prevent the emergence of resistance in (the median OS and RFS had not been reached by a median follow-
patients receiving FLT3-ITD TKI. Other STAT3 inhibitors that work up of 10.9 months).110 A phase I/II study of frontline clofarabine,
as antisense oligonucleotide (ASO) for STAT3 are in clinical trials in cytarabine and idarubicin in patients with intermediate and poor
hematological malignancies including AML and will have to wait risk AML is currently underway (NCT00838240). The potential role
to see their efficacies in near future. of clofarabine in the peri-transplant setting is also currently under
investigation. In a multicenter two-stage phase II trial, 84 patients
IDH1/IDH2 small molecule inhibitors with relapsed and refractory AML received clofarabine (at 30 mg/m2
Gain of function mutations in IDH-1 and IDH-2 enzymes are found for 5 days) in combination with cytarabine as salvage therapy,
in approximately 20% of cases.7 Recent attempts have been followed by 4 days of clofarabine and one dose of melphalan
made to target these mutant enzymes as a potential treatment in chemo-responsive patients with HLA-compatible donors. Out of
for AML. In 2013, Wang et al.102 published the results of AGI-6780, the 56 patients who underwent allo-HSCT, CR was achieved in 50
a small molecule inhibitor of the R140Q mutant IDH-2 enzyme. In an (including 11 CR with incomplete platelet recovery and 10 CR by
ex vivo model of primary human AML cells, treatment with AGI-6780 chimerism). The 2-year OS of 43% compared favorably with historical
was able to overcome the differentiation block of leukemic cells. controls.111 Although randomized controlled trials are needed to
Recently, the IDH-2 inhibitor AG-221 was found to confer a dose- clearly delineate the role of clofarabine in AML these studies present
dependent survival benefits in a primary human IDH-2 mutant promising results in support of this agent, particularly in combina-
AML xenograft model. At a cellular level, AG-221 treatment was tion with cytarabine in the elderly.
associated with an initial phase of CD45+ blast cells proliferation, In addition to its activity as an intravenous agent, clofarabine
followed by cellular differentiation.103 A phase I trial of AG-221 has also captured interest as an oral agent for the treatment of AML.
in IDH-2 mutant leukemia is currently underway (NCT01915498). Unlike previous purine nucleoside analogs, clofarabine is able to
IDH-1 mutant enzymes are also the targets of new therapeutic resist acidic pH as well as phosphorolytic cleavage by gastro-
inhibitors: Preliminary results of a phase I trial of the small molecule intestinal Escherichia coli. As a result it possesses a bioavailability of
inhibitor AG-120 (NCT02074839) demonstrated hematological roughly 50%.112 In a phase I/II study of 35 patients 60 years or older
response in 7 out of 14 IDH-1 positive patients, including 4 CR.104 with relapsed/refractory AML or high-risk MDS the feasibility and
efficacy of oral clofarabine combined with low-dose cytarabine was
Clofarabine investigated as a first-line therapy.113 At a MTD of oral clofarabine
Clofarabine is a second-generation purine nucleoside analog of 20 mg per day for 5 days, CR was achieved in 42% of patients
approved for the treatment of relapsed or refractory pediatric (including CR with incomplete count recovery in 4%). Most
acute lymphocytic leukemia. In AML, it has shown activity and importantly, more than 50% of cycles administered at the MTD
tolerability as a single-agent, administered intravenously at doses of were given in the outpatient setting. In a population at an increased
20–30 mg/m2 for 5 days, with overall response rates of ~ 40%.105,106 risk of TRM, oral clofarabine may thus offer a valuable addition to
In a 2008 randomized study of 70 patients aged 60 years and older, reduced intensity chemotherapy.

Blood Cancer Journal


AML: a comprehensive review and 2016 update
I De Kouchkovsky and M Abdul-Hay
7
Monoclonal antibodies depletion of the normal cells of myeloid lineage is associated
Monoclonal antibodies exert their anti-tumor activity through with unacceptable neutropenia, and attempts to apply CART cell
direct antibody-dependent cytotoxicity or through the conjuga- therapy to the treatment of AML have had to focus on identifying
tion of cytotoxic agents, which allows for the targeted delivery an appropriate antigen to target malignant cells while sparing
of potent chemotherapy to neoplastic cells. Gemtuzumab non-malignant myeloid cells. Kenderian et al. recently developed
ozogamicin (GO) is a humanized recombinant antibody directed a CD33-specific CAR based on the single-chain variable fragment
at CD33, a transmembrane protein expressed on cells of myeloid of gemtuzumab ozogamicin. They have reported potent in vitro
lineage. The antibody is conjugated to the DNA-cleaving cytotoxic activity of such CD33-specific CART cells against AML cell lines.118
agent calicheamicin, and is internalized by CD33-positive cells. GO CD33-specific CART therapy also prolonged survival in AML
received FDA approval in 2000 for the treatment of CD33-positive xenografts. However CD33 is expressed on normal cells of myeloid
AML in patients 60 years or older at first relapse. However in 2009, lineage and the reported anti-tumor effects were associated
interim analysis of a randomized clinical trial of 637 patients with profound cytopenias. Using electroporation of CD33-specific
with newly diagnosed AML revealed increased fatal toxicity with
CAR mRNA into human T cells, the authors are able to induce the
no improvement in CR, disease-free survival or OS in patients
transient expression of anti-CD33 CAR,118 which may confer
receiving GO in combination with standard chemotherapy.114 The
trial was prematurely terminated and FDA approval was rescinded. clinically significant anti-tumor activity while avoiding long-term
Despite its removal from the market, a 2014 meta-analysis of five myelosuppression. In an attempt to selectively target leukemic
randomized clinical trials demonstrated a decrease in relapse myeloid cells, others have focused instead on the β member of the
and improved survival in patients receiving GO in addition to folate receptor family (FRβ). This receptor subtype is primarily
standard chemotherapy.115 A subset analysis further revealed expressed on myeloid-lineage hematopoietic cells, and is upregu-
that this survival benefit was limited to patients with a favorable or lated in the setting of malignancy.119 FRβ is expressed in 70% of
intermediate cytogenetic-risk profile. Most recently, a randomized cases of primary AML, and its expression can be further upregulated
trial of 237 patients 60 years of age or older ineligible for intensive following treatment with all-trans retinoic acid.120 In a recent
chemotherapy showed an improved OS (hazard ratio of 0.69) in publication the effect of FRβ-specific CART cells therapy in vitro as
those assigned to GO induction and consolidation compared well as in AML xenograft was reported. The study demonstrated lytic
with best supportive care.116 This survival benefit was again activity against FRβ positive AML cell lines both in vitro and in vivo.
most pronounced in patients with an intermediate to favorable Importantly, no evidence of toxic activity against healthy human
cytogenetic-risk profile. Although further studies are required CD34-positive stem cells was observed in vitro.121 Although still in its
to fully delineate the effects of gemtuzumab in the treatment early stages, CART cell therapy may thus provide an alternative
of AML, these data suggest a benefit among elderly patients mechanism of treatment for patients with relapsed/refractory AML.
with favorable or intermediate cytogenetic-risk profile. CD37 is
a transmembrane protein that is expressed in high levels on
maturing B cells. Although its exact function has not yet been
elucidated, it is upregulated in non-Hogkin lymphoma and chronic CONCLUSION
lymphocytic leukemia.117 Initially conceived as a therapy for B-cell AML is a biologically and clinically heterogeneous disease. Although
malignancies, AGS67E is a fully human anti-CD37 IgG antibody advances in supportive care and prognostic risk stratification
that is conjugated with monomethyl auristatin E (MMAE), a potent have optimized established therapies, overall long-term survival
microtubule-disrupting agent. AGS67E allows for the selective remains poor. Elderly patients who account for the majority of
delivery of MMAE to CD37-positive malignant cells and results
newly diagnosed cases are more likely to present with an adverse
in apoptosis. Although CD37 is minimally expressed on normal
cytogenetic profile. At the same time the increased risk of TRM often
myeloid stem cells it was recently demonstrated to have differential
expression of CD37 on the surface of CD34+/CD38− AML stem cells. precludes this population of patients from receiving optimal
In vitro treatment of leukemic cells with nanomolar concentrations chemotherapy or stem cell transplantation. Novel targeted therapies
of AGS67E resulted in cytotoxicity, altered cell growth and apoptosis offer the promise of effective anti-leukemic activity with reduced
in seven out of 16 AML cell lines. The administration of AGS67E was toxicity from off-target effects. However given the molecular
further found to significantly decreased tumor engraftment in diversity of AML, it is unlikely that targeted therapies such as FLT3
a murine xenograft model of AML, resulting in undetectable tyrosine kinase inhibitors will provide a single ‘magic bullet’ against
leukemic cell levels in three out of four AML samples. CD37 may this disease. Rather the development of new treatments, in concert
thus represent a novel therapeutic target for the selective inhibition with improved genetic profiling and risk stratification, can be
of leukemic cell growth. Although still at an early stage of its clinical expected to result in incremental gains in remission and survival.
development, AGS67E is a promising new therapy. More impor- Furthermore in addition to mutated enzymes and upregulated
tantly, the identification of unique molecular markers expressed on pathways, the identification of unique cell surface markers can
the surface of leukemic cells is an emerging avenue for the provide a therapeutic target for recombinant monoclonal antibodies
discovery of novel targeted therapies against AML. or chimeric antigen receptors. Here, the challenge lays in selectively
targeting leukemic myeloid cells while sparing non-malignant
CART therapy myeloid precursors. Lastly, the development of well-tolerated oral
Chimeric antigen receptors are synthetic T-cell receptors with therapies, such as clofarabine, will increasingly broaden the range of
antibody-like specificity. They combine a single-chain variable available treatment for elderly patients at a higher risk of mortality
fragment from a monoclonal antibody with the transmembrane from standard chemotherapy regimens. We are looking to a new era
and intracellular domains of a T-cell receptor. This allows for the in the treatment of AML to begin with novel agents so we can
creation of a host-derived population of chimeric antigen receptor-T achieve better responses with prolong OS particularly for patients
(CART) cells, which can be directed at a pre-determined antigen. with relapsed or refractory diseases and poor cytogenetic features.
CD19-directed CART cell therapy has shown exciting efficacy in the
treatment of acute lymphoblastic leukemia and B-cell lymphoma.
Although this treatment does not distinguish between malignant
and healthy CD19 cells, patients tolerate the depletion of CD19 CONFLICT OF INTEREST
lymphocytes with relatively little morbidity. In contrast to this, The authors declare no conflict of interest.

Blood Cancer Journal


AML: a comprehensive review and 2016 update
I De Kouchkovsky and M Abdul-Hay
8
REFERENCES leukemia: a comprehensive large-scale study from a single Chinese center.
1 Yamamoto JF, Goodman MT. Patterns of leukemia incidence in the United States Leuk Res 2014; 38: 1435–1440.
by subtype and demographic characteristics, 1997-2002. Cancer Causes Control 26 Paschka P, Marcucci G, Ruppert AS, Mrozek K, Chen H, Kittles RA et al. Adverse
2008; 19: 379–390. prognostic significance of KIT mutations in adult acute myeloid leukemia with
2 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin 2015; 65:. 5–29. inv(16) and t(8;21): a Cancer and Leukemia Group B Study. J Clin Oncol 2006; 24:
3 Shah A, Andersson TM, Rachet B, Bjorkholm M, Lambert PC. Survival and cure 3904–3911.
of acute myeloid leukaemia in England, 1971-2006: a population-based study. 27 Park SH, Chi HS, Min SK, Park BG, Jang S, Park CJ. Prognostic impact of c-KIT mutations
Br J Haematol 2013; 162: 509–516. in core binding factor acute myeloid leukemia. Leuk Res 2011; 35: 1376–1383.
4 Meyers J, Yu Y, Kaye JA, Davis KL. Medicare fee-for-service enrollees with primary 28 Park SH, Lee HJ, Kim IS, Kang JE, Lee EY, Kim HJ et al. Incidences and prognostic
acute myeloid leukemia: an analysis of treatment patterns, survival, and healthcare impact of c-KIT, WT1, CEBPA, and CBL mutations, and mutations associated
resource utilization and costs. Appl Health Econ Health Policy 2013; 11: 275–286. with epigenetic modification in core binding factor acute myeloid leukemia:
5 Sill H, Olipitz W, Zebisch A, Schulz E, Wolfler A. Therapy-related myeloid a multicenter study in a Korean population. Ann Lab Med 2015; 35: 288–297.
neoplasms: pathobiology and clinical characteristics. Br J Pharmacol 2011; 162; 29 Dohner K, Schlenk RF, Habdank M, Scholl C, Rucker FG, Corbacioglu A et al.
792–805. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults
6 Patel JP, Gonen M, Figueroa ME, Fernandez H, Sun Z, Racevskis J et al. Prognostic with acute myeloid leukemia and normal cytogenetics: interaction with other
relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med gene mutations. Blood 2005; 106: 3740–3746.
2012; 366: 1079–1089. 30 Li HY, Deng DH, Huang Y, Ye FH, Huang LL, Xiao Q et al. Favorable prognosis
7 Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of biallelic CEBPA gene mutations in acute myeloid leukemia patients:
a meta-analysis. Eur J Haematol 2015; 94: 439–448.
of adult de novo acute myeloid leukemia. N Engl J Med 2013; 368: 2059–2074.
31 Frohling S, Schlenk RF, Breitruck J, Benner A, Kreitmeier S, Tobis K et al. Prog-
8 Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood
nostic significance of activating FLT3 mutations in younger adults (16 to 60
2002; 100: 1532–1542.
years) with acute myeloid leukemia and normal cytogenetics: a study of the AML
9 Takahashi S. Current findings for recurring mutations in acute myeloid leukemia.
Study Group Ulm. Blood 2002; 100: 4372–4380.
J Hematol Oncol 2011; 4: 36.
32 Port M, Bottcher M, Thol F, Ganser A, Schlenk R, Wasem J et al. Prognostic
10 Kihara R, Nagata Y, Kiyoi H, Kato T, Yamamoto E, Suzuki K et al. Comprehensive
significance of FLT3 internal tandem duplication, nucleophosmin 1, and CEBPA
analysis of genetic alterations and their prognostic impacts in adult acute
gene mutations for acute myeloid leukemia patients with normal karyotype and
myeloid leukemia patients. Leukemia 2014; 28: 1586–1595.
younger than 60 years: a systematic review and meta-analysis. Ann Hematol
11 Cook AM, Li L, Ho Y, Lin A, Li L, Stein A et al. Role of altered growth factor
2014; 93: 1279–1286.
receptor-mediated JAK2 signaling in growth and maintenance of human acute
33 Cagnetta A, Adamia S, Acharya C, Patrone F, Miglino M, Nencioni A et al. Role of
myeloid leukemia stem cells. Blood 2014; 123: 2826–2837.
genotype-based approach in the clinical management of adult acute myeloid
12 Ghoshal Gupta S, Baumann H, Wetzler M. Epigenetic regulation of signal
leukemia with normal cytogenetics. Leuk Res 2014; 38: 649–659.
transducer and activator of transcription 3 in acute myeloid leukemia. Leuk Res
34 Gale RE, Green C, Allen C, Mead AJ, Burnett AK, Hills RK et al. The impact of FLT3
2008; 32: 1005–1014.
internal tandem duplication mutant level, number, size, and interaction with
13 Yamada O, Kawauchi K. The role of the JAK-STAT pathway and related signal
NPM1 mutations in a large cohort of young adult patients with acute myeloid
cascades in telomerase activation during the development of hematologic
leukemia. Blood 2008; 111: 2776–2784.
malignancies. JAKSTAT 2013; 2: e25256.
35 Schnittger S, Bacher U, Haferlach C, Alpermann T, Kern W, Haferlach T. Diversity
14 Schuringa JJ, Wierenga AT, Kruijer W, Vellenga E. Constitutive Stat3, Tyr705, and
of the juxtamembrane and TKD1 mutations (exons 13-15) in the FLT3 gene
Ser727 phosphorylation in acute myeloid leukemia cells caused by the autocrine
with regards to mutant load, sequence, length, localization, and correlation with
secretion of interleukin-6. Blood 2000; 95: 3765–3770.
biological data. Genes Chromosomes Cancer 2012; 51: 910–924.
15 Spiekermann K, Bagrintseva K, Schwab R, Schmieja K, Hiddemann W. Over-
36 Grossmann V, Schnittger S, Kohlmann A, Eder C, Roller A, Dicker F et al. A novel
expression and constitutive activation of FLT3 induces STAT5 activation in pri-
hierarchical prognostic model of AML solely based on molecular mutations.
mary acute myeloid leukemia blast cells. Clin Cancer Res 2003; 9: 2140–2150. Blood 2012; 120: 2963–2972.
16 Steensma DP, McClure RF, Karp JE, Tefferi A, Lasho TL, Powell HL et al. JAK2 37 Shivarov V, Gueorguieva R, Stoimenov A, Tiu R. DNMT3A mutation is a poor
V617F is a rare finding in de novo acute myeloid leukemia, but STAT3 activation prognosis biomarker in AML: results of a meta-analysis of 4500 AML patients.
is common and remains unexplained. Leukemia 2006; 20: 971–978. Leuk Res 2013; 37: 1445–1450.
17 Dohner H, Estey EH, Amadori S, Appelbaum FR, Buchner T, Burnett AK et al. 38 DiNardo CD, Ravandi F, Agresta S, Konopleva M, Takahashi K, Kadia T et al.
Diagnosis and management of acute myeloid leukemia in adults: recommen- Characteristics, clinical outcome, and prognostic significance of IDH mutations in
dations from an international expert panel, on behalf of the European Leuke- AML. Am J Hematol 2015; 90: 732–736.
miaNet. Blood 2010; 115: 453–474. 39 Chen X, Xie H, Wood BL, Walter RB, Pagel JM, Becker PS et al. Relation of clinical
18 Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A et al. The 2008 response and minimal residual disease and their prognostic impact on outcome
revision of the World Health Organization (WHO) classification of myeloid neoplasms in acute myeloid leukemia. J Clin Oncol 2015; 33: 1258–1264.
and acute leukemia: rationale and important changes. Blood 2009; 114:. 937–951. 40 Walter RB, Kantarjian HM, Huang X, Pierce SA, Sun Z, Gundacker HM et al. Effect
19 Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM et al. The 2016 of complete remission and responses less than complete remission on survival
revision to the World Health Organization (WHO) classification of myeloid in acute myeloid leukemia: a combined Eastern Cooperative Oncology Group,
neoplasms and acute leukemia. Blood 2016; 127: 2391–2405. Southwest Oncology Group, and M. D. Anderson Cancer Center Study. J Clin
20 Kantarjian H, O'Brien S, Cortes J, Giles F, Faderl S, Jabbour E et al. Results of Oncol 2010; 28: 1766–1771.
intensive chemotherapy in 998 patients age 65 years or older with acute mye- 41 Hoyos M, Nomdedeu JF, Esteve J, Duarte R, Ribera JM, Llorente A et al. Core binding
loid leukemia or high-risk myelodysplastic syndrome: predictive prognostic factor acute myeloid leukemia: the impact of age, leukocyte count, molecular
models for outcome. Cancer 2006; 106: 1090–1098. findings, and minimal residual disease. Eur J Haematol 2013; 91: 209–218.
21 Walter RB, Othus M, Borthakur G, Ravandi F, Cortes JE, Pierce SA et al. Prediction 42 Yin JA, O'Brien MA, Hills RK, Daly SB, Wheatley K, Burnett AK. Minimal residual
of early death after induction therapy for newly diagnosed acute myeloid disease monitoring by quantitative RT-PCR in core binding factor AML allows risk
leukemia with pretreatment risk scores: a novel paradigm for treatment stratification and predicts relapse: results of the United Kingdom MRC
assignment. J Clin Oncol 2011; 29: 4417–4423. AML-15 trial. Blood 2012; 120: 2826–2835.
22 Hulegardh E, Nilsson C, Lazarevic V, Garelius H, Antunovic P, Derolf A. Rangert 43 Buccisano F, Maurillo L, Del Principe MI, Del Poeta G, Sconocchia G, Lo-Coco F
et al. Characterization and prognostic features of secondary acute myeloid et al. Prognostic and therapeutic implications of minimal residual disease
leukemia in a population-based setting: a report from the Swedish Acute Leu- detection in acute myeloid leukemia. Blood 2012; 119: 332–341.
kemia Registry. Am J Hematol 2015; 90: 208–214. 44 Buckley SA, Appelbaum FR, Walter RB. Prognostic and therapeutic implications
23 Estey EH. Acute myeloid leukemia: 2014 update on risk-stratification and man- of minimal residual disease at the time of transplantation in acute leukemia.
agement. Am J Hematol 2014; 89: 1063–1081. Bone Marrow Transplant 2013; 48: 630–641.
24 Mrozek K, Marcucci G, Nicolet D, Maharry KS, Becker H, Whitman SP et al. 45 Vidriales MB, Perez-Lopez E, Pegenaute C, Castellanos M, Perez JJ, Chandia M
Prognostic significance of the European LeukemiaNet standardized system for et al. Minimal residual disease evaluation by flow cytometry is a complementary
reporting cytogenetic and molecular alterations in adults with acute myeloid tool to cytogenetics for treatment decisions in acute myeloid leukaemia. Leuk
leukemia. J Clin Oncol 2012; 30: 4515–4523. Res 2015; 40: 1–9.
25 Qin YZ, Zhu HH, Jiang Q, Jiang H, Zhang LP, Xu LP et al. Prevalence and prog- 46 Gong Q, Zhou L, Xu S, Li X, Zou Y, Chen J. High doses of daunorubicin during
nostic significance of c-KIT mutations in core binding factor acute myeloid induction therapy of newly diagnosed acute myeloid leukemia: a systematic

Blood Cancer Journal


AML: a comprehensive review and 2016 update
I De Kouchkovsky and M Abdul-Hay
9
review and meta-analysis of prospective clinical trials. PLoS One 2015; 10: 67 Cornelissen JJ, van Putten WL, Verdonck LF, Theobald M, Jacky E, Daenen SM
e0125612. et al. Results of a HOVON/SAKK donor versus no-donor analysis of myeloablative
47 Li X, Xu S, Tan Y, Chen J. The effects of idarubicin versus other anthracyclines HLA-identical sibling stem cell transplantation in first remission acute myeloid
for induction therapy of patients with newly diagnosed leukaemia. Cochrane leukemia in young and middle-aged adults: benefits for whom? Blood 2007; 109:
Database Syst Rev 2015; 6: CD010432. 3658–3666.
48 Lowenberg B. Sense and nonsense of high-dose cytarabine for acute myeloid 68 Schetelig J, Schaich M, Schafer-Eckart K, Hanel M, Aulitzky WE, Einsele H et al.
leukemia. Blood 2013; 121: 26–28. Hematopoietic cell transplantation in patients with intermediate and high-risk
49 Lowenberg B, Pabst T, Vellenga E, van Putten W, Schouten HC, Graux C et al. AML: results from the randomized Study Alliance Leukemia (SAL) AML 2003 trial.
Cytarabine dose for acute myeloid leukemia. N Engl J Med 2011; 364: 1027–1036. Leukemia 2015; 29:. 1060–1068.
50 Burnett AK, Russell NH, Hills RK, Hunter AE, Kjeldsen L, Yin J et al. Optimization of 69 Li D, Wang L, Zhu H, Dou L, Liu D, Fu L et al. Efficacy of allogeneic hematopoietic
chemotherapy for younger patients with acute myeloid leukemia: results of the stem cell transplantation in intermediate-risk acute myeloid leukemia
medical research council AML15 trial. J Clin Oncol 2013; 31: 3360–3368. adult patients in first complete remission: a meta-analysis of prospective studies.
51 Lowenberg B, Zittoun R, Kerkhofs H, Jehn U, Abels J, Debusscher L et al. On the PLoS One 2015; 10: e0132620.
value of intensive remission-induction chemotherapy in elderly patients of 70 Schlenk RF, Kayser S, Bullinger L, Kobbe G, Casper J, Ringhoffer M et al.
65+ years with acute myeloid leukemia: a randomized phase III study of the Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with
European Organization for Research and Treatment of Cancer Leukemia Group. respect to allogeneic transplantation. Blood 2014; 124: 3441–3449.
J Clin Oncol 1989; 7: 1268–1274. 71 Zhang W, Konopleva M, Shi YX, McQueen T, Harris D, Ling X et al. Mutant FLT3:
52 Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Gattermann N, Germing U a direct target of sorafenib in acute myelogenous leukemia. J Natl Cancer Inst
et al. Azacitidine prolongs overall survival compared with conventional care 2008; 100: 184–198.
regimens in elderly patients with low bone marrow blast count acute myeloid 72 Borthakur G, Kantarjian H, Ravandi F, Zhang W, Konopleva M, Wright JJ et al.
leukemia. J Clin Oncol 2010; 28: 562–569. Phase I study of sorafenib in patients with refractory or relapsed acute leuke-
53 Kantarjian HM, Thomas XG, Dmoszynska A, Wierzbowska A, Mazur G, Mayer J mias. Haematologica 2011; 96: 62–68.
et al. Multicenter, randomized, open-label, phase III trial of decitabine versus 73 Crump M, Hedley D, Kamel-Reid S, Leber B, Wells R, Brandwein J et al.
patient choice, with physician advice, of either supportive care or low-dose A randomized phase I clinical and biologic study of two schedules of sorafenib in
cytarabine for the treatment of older patients with newly diagnosed acute patients with myelodysplastic syndrome or acute myeloid leukemia: a NCIC
myeloid leukemia. J Clin Oncol 2012; 30: 2670–2677. (National Cancer Institute of Canada) Clinical Trials Group Study. Leuk Lymphoma
54 Dombret H, Seymour JF, Butrym A, Wierzbowska A, Selleslag D, Jang JH et al. 2010; 51: 252–260.
International phase 3 study of azacitidine vs conventional care regimens in older 74 Pratz KW, Cho E, Levis MJ, Karp JE, Gore SD, McDevitt M et al. A pharmacody-
patients with newly diagnosed AML with 430% blasts. Blood 2015; 126: namic study of sorafenib in patients with relapsed and refractory acute leuke-
291–299. mias. Leukemia 2010; 24: 1437–1444.
55 Fernandez HF, Sun Z, Yao X, Litzow MR, Luger SM, Paietta EM et al. Anthracycline 75 Al-Kali A, Cortes J, Faderl S, Jones D, Abril C, Pierce S et al. Patterns of molecular
dose intensification in acute myeloid leukemia. N Engl J Med 2009; 361: response to and relapse after combination of sorafenib, idarubicin, and
1249–1259. cytarabine in patients with FLT3 mutant acute myeloid leukemia. Clin Lymphoma
56 Carella AM, Cascavilla N, Greco MM, Melillo L, Sajeva MR, Ladogana S et al. Myeloma Leuk 2011; 11: 361–366.
Treatment of "poor risk" acute myeloid leukemia with fludarabine, cytarabine and 76 Man CH, Fung TK, Ho C, Han HH, Chow HC, Ma AC et al. Sorafenib treatment of
G-CSF (flag regimen): a single center study. Leuk Lymphoma 2001; 40: 295–303. FLT3-ITD(+) acute myeloid leukemia: favorable initial outcome and mechanisms
57 Herzig RH, Lazarus HM, Wolff SN, Phillips GL, Herzig GP. High-dose cytosine ara- of subsequent nonresponsiveness associated with the emergence of a D835
binoside therapy with and without anthracycline antibiotics for remission rein- mutation. Blood 2012; 119: 5133–5143.
duction of acute nonlymphoblastic leukemia. J Clin Oncol 1985; 3: 992–997. 77 Ravandi F, Cortes JE, Jones D, Faderl S, Garcia-Manero G, Konopleva MY et al.
58 Amadori S, Arcese W, Isacchi G, Meloni G, Petti MC, Monarca B et al. Mitoxan- Phase I/II study of combination therapy with sorafenib, idarubicin, and
trone, etoposide, and intermediate-dose cytarabine: an effective and tolerable cytarabine in younger patients with acute myeloid leukemia. J Clin Oncol 2010;
regimen for the treatment of refractory acute myeloid leukemia. J Clin Oncol 28: 1856–1862.
1991; 9: 1210–1214. 78 Konig H, Levis M. Targeting FLT3 to treat leukemia. Expert Opin Ther Targets 2015;
59 Daenen S, Lowenberg B, Sonneveld P, van Putten WL, Verhoef G, Verdonck LF 19: 37–54.
et al. Efficacy of etoposide and mitoxantrone in patients with acute myelo- 79 Ravandi F, Alattar ML, Grunwald MR, Rudek MA, Rajkhowa T, Richie MA et al.
genous leukemia refractory to standard induction therapy and intermediate- Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid
dose cytarabine with amsidine. Dutch Hematology-Oncology Working Group for leukemia and FLT-3 internal tandem duplication mutation. Blood 2013; 121:
Adults (HOVON). Leukemia 1994; 8: 6–10. 4655–4662.
60 Buchner T, Schlenk RF, Schaich M, Dohner K, Krahl R, Krauter J et al. Acute 80 Serve H, Krug U, Wagner R, Sauerland MC, Heinecke A, Brunnberg U et al.
myeloid leukemia (AML): different treatment strategies versus a common Sorafenib in combination with intensive chemotherapy in elderly patients
standard arm--combined prospective analysis by the German AML Intergroup. with acute myeloid leukemia: results from a randomized, placebo-
J Clin Oncol 2012; 30: 3604–3610. controlled trial. J Clin Oncol 2013; 31: 3110–3118.
61 Koreth J, Schlenk R, Kopecky KJ, Honda S, Sierra J, Djulbegovic BJ et al. 81 Rollig C, Serve H, Huttmann A, Noppeney R, Muller-Tidow C, Krug U et al.
Allogeneic stem cell transplantation for acute myeloid leukemia in first complete Addition of sorafenib versus placebo to standard therapy in patients aged 60
remission: systematic review and meta-analysis of prospective clinical trials. years or younger with newly diagnosed acute myeloid leukaemia (SORAML):
JAMA 2009; 301: 2349–2361. a multicentre, phase 2, randomised controlled trial. Lancet Oncol 2015; 16;
62 Yanada M, Matsuo K, Emi N, Naoe T. Efficacy of allogeneic hematopoietic stem 1691–1699.
cell transplantation depends on cytogenetic risk for acute myeloid leukemia in 82 Sato T, Yang X, Knapper S, White P, Smith BD, Galkin S et al. FLT3 ligand impedes
first disease remission: a metaanalysis. Cancer 2005; 103:. 1652–1658. the efficacy of FLT3 inhibitors in vitro and in vivo. Blood 2011; 117: 3286–3293.
63 Weick JK, Kopecky KJ, Appelbaum FR, Head DR, Kingsbury LL, Balcerzak SP et al. 83 Chen YB, Li S, Lane AA, Connolly C, Del Rio C, Valles B et al. Phase I trial of
A randomized investigation of high-dose versus standard-dose cytosine arabi- maintenance sorafenib after allogeneic hematopoietic stem cell transplantation
noside with daunorubicin in patients with previously untreated acute myeloid for fms-like tyrosine kinase 3 internal tandem duplication acute myeloid leu-
leukemia: a Southwest Oncology Group study. Blood 1996; 88: 2841–2851. kemia. Biol Blood Marrow Transplant 2014; 20: 2042–2048.
64 Stelljes M, Krug U, Beelen DW, Braess J, Sauerland MC, Heinecke A et al. 84 Antar A, Kharfan-Dabaja MA, Mahfouz R, Bazarbachi A. Sorafenib maintenance
Allogeneic transplantation versus chemotherapy as postremission therapy for appears safe and improves clinical outcomes in FLT3-ITD acute myeloid leuke-
acute myeloid leukemia: a prospective matched pairs analysis. J Clin Oncol 2014; mia after allogeneic hematopoietic cell transplantation. Clin Lymphoma Myeloma
32: 288–296. Leuk 2015; 15: 298–302.
65 Schlenk RF, Dohner K, Krauter J, Frohling S, Corbacioglu A, Bullinger L et al. 85 Metzelder SK, Schroeder T, Finck A, Scholl S, Fey M, Gotze K et al. High activity of
Mutations and treatment outcome in cytogenetically normal acute myeloid sorafenib in FLT3-ITD-positive acute myeloid leukemia synergizes with allo-
leukemia. N Engl J Med 2008; 358: 1909–1918. immune effects to induce sustained responses. Leukemia 2012; 26: 2353–2359.
66 Rollig C, Bornhauser M, Kramer M, Thiede C, Ho AD, Kramer A et al. Allogeneic 86 Fischer T, Stone RM, Deangelo DJ, Galinsky I, Estey E, Lanza C et al. Phase IIB trial
stem-cell transplantation in patients with NPM1-mutated acute myeloid of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and
leukemia: results from a prospective donor versus no-donor analysis of patients multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and
after upfront HLA typing within the SAL-AML 2003 trial. J Clin Oncol 2015; 33; high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. J Clin
403–410. Oncol 2010; 28: 4339–4345.

Blood Cancer Journal


AML: a comprehensive review and 2016 update
I De Kouchkovsky and M Abdul-Hay
10
87 Stone RM, DeAngelo DJ, Klimek V, Galinsky I, Estey E, Nimer SD et al. Patients 106 Kantarjian HM, Erba HP, Claxton D, Arellano M, Lyons RM, Kovascovics T et al.
with acute myeloid leukemia and an activating mutation in FLT3 respond to Phase II study of clofarabine monotherapy in previously untreated older adults
a small-molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood 2005; 105: 54–60. with acute myeloid leukemia and unfavorable prognostic factors. J Clin Oncol
88 Cooper BW, Kindwall-Keller TL, Craig MD, Creger RJ, Hamadani M, Tse WW et al. 2010; 28: 549–555.
A phase I study of midostaurin and azacitidine in relapsed and elderly AML 107 Faderl S, Ravandi F, Huang X, Garcia-Manero G, Ferrajoli A, Estrov Z et al.
patients. Clin Lymphoma Myeloma Leuk 2015; 15: 428–432, e2. A randomized study of clofarabine versus clofarabine plus low-dose cytarabine
89 Strati P, Kantarjian H, Ravandi F, Nazha A, Borthakur G, Daver N et al. Phase I/II as front-line therapy for patients aged 60 years and older with acute myeloid
trial of the combination of midostaurin (PKC412) and 5-azacytidine for patients leukemia and high-risk myelodysplastic syndrome. Blood 2008; 112: 1638–1645.
with acute myeloid leukemia and myelodysplastic syndrome. Am J Hematol 108 Faderl S, Wetzler M, Rizzieri D, Schiller G, Jagasia M, Stuart R et al. Clofarabine
2015; 90: 276–281. plus cytarabine compared with cytarabine alone in older patients with relapsed
90 Stone RM, Mandrekar S, Sanford BL, Geyer S, Bloomfield CD, Dohner K et al. or refractory acute myelogenous leukemia: results from the CLASSIC I Trial. J Clin
57th Annual Meeting & Exposition The Multi-Kinase Inhibitor Midostaurin (M) Oncol 2012; 30: 2492–2499.
Prolongs Survival Compared with Placebo (P) in Combination with Daunorubicin 109 Kadia TM, Faderl S, Ravandi F, Jabbour E, Garcia-Manero G, Borthakur G et al.
(D)/Cytarabine (C) Induction (ind), High-Dose C Consolidation (consol), and As Final results of a phase 2 trial of clofarabine and low-dose cytarabine alternating
Maintenance (maint) Therapy in Newly Diagnosed Acute Myeloid Leukemia with decitabine in older patients with newly diagnosed acute myeloid leukemia.
(AML) Patients (pts) Age 18-60 with FLT3 Mutations (muts): An International Cancer 2015; 121: 2375–2382.
Prospective Randomized (rand) P-Controlled Double-Blind Trial.Am Soc Hematol 110 Nazha A, Kantarjian H, Ravandi F, Huang X, Choi S, Garcia-Manero G et al.
2015, Orlando, FL, USA. 5–8 December 2015. Clofarabine, idarubicin, and cytarabine (CIA) as frontline therapy for patients
91 Cortes JE, Kantarjian H, Foran JM, Ghirdaladze D, Zodelava M, Borthakur G et al. p 60 years with newly diagnosed acute myeloid leukemia. Am J Hematol 2013;
Phase I study of quizartinib administered daily to patients with relapsed 88: 961–966.
or refractory acute myeloid leukemia irrespective of FMS-like tyrosine kinase 111 Middeke JM, Herbst R, Parmentier S, Bug G, Hanel M, Stuhler G et al. Clofarabine
3-internal tandem duplication status. J Clin Oncol 2013; 31: 3681–3687. salvage therapy before allogeneic hematopoietic stem cell transplantation in
92 Levis M, Ravandi F, Wang ES, Baer MR, Perl A, Coutre S et al. Results from a patients with relapsed or refractory AML: results of the BRIDGE trial. Leukemia
randomized trial of salvage chemotherapy followed by lestaurtinib for patients 2015; 30: 261–267.
with FLT3 mutant AML in first relapse. Blood 2011; 117: 3294–3301. 112 Bonate PL, Cunningham CC, Gaynon P, Jeha S, Kadota R, Lam GN et al.
93 Levis MJ, Perl AE, Dombret H, Döhner H, Steffen B, Rousselot P et al. Final results Population pharmacokinetics of clofarabine and its metabolite 6-ketoclofarabine
of a phase 2 Open-label, monotherapy efficacy and safety study of quizartinib in adult and pediatric patients with cancer. Cancer Chemother Pharmacol 2011;
(AC220) in patients with FLT3-ITD positive or negative relapsed/refractory 67: 875–890.
acute myeloid leukemia after second-line chemotherapy or hematopoietic stem 113 Buckley SA, Mawad R, Gooley TA, Becker PS, Sandhu V, Hendrie P et al. A phase I/
cell transplantation. Blood 2012, 120: 673. ASH abstract 673. II study of oral clofarabine plus low-dose cytarabine in previously treated acute
94 Borthakur G, Kantarjian HM, O'Brien S, Garcia-Manero G, Jabbour E, Daver N et al. myeloid leukaemia and high-risk myelodysplastic syndrome patients at least 60
The combination of Quizartinib with Azacitidine or low dose Cytarabine is highly years of age. Br J Haematol 2015; 170: 349–355.
active in patients (Pts) with FLT3-ITD mutated myeloid leukemias: interim report 114 Petersdorf SH, Kopecky KJ, Slovak M, Willman C, Nevill T, Brandwein J et al.
of a phase I/II trial. Blood 2014; 124: 388–388. A phase 3 study of gemtuzumab ozogamicin during induction and post-
95 Smith CC, Lasater EA, Lin KC, Wang Q, McCreery MQ, Stewart WK et al. consolidation therapy in younger patients with acute myeloid leukemia. Blood
Crenolanib is a selective type I pan-FLT3 inhibitor. Proc Natl Acad Sci USA 2014; 2013; 121: 4854–4860.
111: 5319–5324. 115 Hills RK, Castaigne S, Appelbaum FR, Delaunay J, Petersdorf S, Othus M et al.
96 Randhawa JK, Kantarjian H, Borthakur G, Thompson PA, Konopleva M, Daver N Addition of gemtuzumab ozogamicin to induction chemotherapy in adult
et al. 57th Annual Meeting & Exposition. Results of a phase II study of crenolanib patients with acute myeloid leukaemia: a meta-analysis of individual patient
in relapsed/refractory acute myeloid leukemia patients (Pts) with activating FLT3 data from randomised controlled trials. Lancet Oncol 2014; 15: 986–996.
mutations. Am Soc Hematol 2014, San Francisco, CA, USA. 116 Amadori S, Suciu S, Selleslag D, Aversa F, Gaidano G, Musso M et al. Gemtuzu-
97 Zhou J, Bi C, Janakakumara JV, Liu SC, Chng WJ, Tay KG et al. Enhanced mab Ozogamicin versus best supportive care in older patients with newly
activation of STAT pathways and overexpression of survivin confer resistance diagnosed acute myeloid leukemia unsuitable for intensive chemotherapy:
to FLT3 inhibitors and could be therapeutic targets in AML. Blood 2009; 113: results of the randomized phase III EORTC-GIMEMA AML-19 Trial. J Clin Oncol
4052–4062. 2016; 34: 972–979.
98 Redell MS, Ruiz MJ, Alonzo TA, Gerbing RB, Tweardy DJ. Stat3 signaling in acute 117 Moore K, Cooper SA, Jones DB. Use of the monoclonal antibody WR17,
myeloid leukemia: ligand-dependent and -independent activation and induction identifying the CD37 gp40-45 Kd antigen complex, in the diagnosis of B-lym-
of apoptosis by a novel small-molecule Stat3 inhibitor. Blood 2011; 117: phoid malignancy. J Pathol 1987; 152: 13–21.
5701–5709. 118 Kenderian SS, Ruella M, Shestova O, Klichinsky M, Aikawa V, Morrissette JJ et al.
99 Krueger MJ, Minus M, Liu W, Long X, Stevens AM, Kolosov MI et al. 57th Annual CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity
Meeting & Exposition. A novel STAT3 inhibitor has potent activity in preclinical against human acute myeloid leukemia. Leukemia 2015; 29: 1637–1647.
models of acute myeloid leukemia that incorporate the stromal environment. 119 Ross JF, Wang H, Behm FG, Mathew P, Wu M, Booth R et al. Folate receptor type
Am Soc Hematol 2015, Orlando, FL, USA. beta is a neutrophilic lineage marker and is differentially expressed in myeloid
100 Oh DY, Lee SH, Han SW, Kim MJ, Kim TM, Kim TY et al. Phase I study of leukemia. Cancer 1999; 85: 348–357.
OPB-31121, an oral STAT3 inhibitor, in patients with advanced solid tumors. 120 Wang H, Zheng X, Behm FG, Ratnam M. Differentiation-independent retinoid
Cancer Res Treat 2015; 47: 607–615. induction of folate receptor type beta, a potential tumor target in myeloid
101 Hayakawa F, Sugimoto K, Harada Y, Hashimoto N, Ohi N, Kurahashi S et al. leukemia. Blood 2000; 96: 3529–3536.
A novel STAT inhibitor, OPB-31121, has a significant antitumor effect on 121 Lynn RC, Poussin M, Kalota A, Feng Y, Low PS, Dimitrov DS et al. Targeting of
leukemia with STAT-addictive oncokinases. Blood Cancer J 2013; 3: e166. folate receptor beta on acute myeloid leukemia blasts with chimeric antigen
102 Wang F, Travins J, DeLaBarre B, Penard-Lacronique V, Schalm S, Hansen E et al. receptor-expressing T cells. Blood 2015; 125: 3466–3476.
Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differ-
entiation. Science 2013; 340: 622–626.
103 Ellwood-Yen K, Wang F, Travins J, Chen Y, Yang H, Straley K et al. AG-221 offers This work is licensed under a Creative Commons Attribution 4.0
a survival advantage in a primary human IDH2 mutant AML xenograft model. International License. The images or other third party material in this
in AACR Annual Meeting. 2014. San Diego, CA. article are included in the article’s Creative Commons license, unless indicated
104 Pollyea DA, Botton Sd, Fathi AT, Stein EM, Tallman MS, Agresta S et al. Clinical otherwise in the credit line; if the material is not included under the Creative Commons
safety and activity in a phase I trial of AG-120, a first in class, selective, potent license, users will need to obtain permission from the license holder to reproduce the
inhibitor of the IDH1-mutant protein, in patients with IDH1 mutant positive material. To view a copy of this license, visit http://creativecommons.org/licenses/
advanced hematologic malignancies. Eur J Cancer 2014; 50: 195. by/4.0/
105 Burnett AK, Russell NH, Hunter AE, Milligan D, Knapper S, Wheatley K et al.
Clofarabine doubles the response rate in older patients with acute myeloid
leukemia but does not improve survival. Blood 2013; 122: 1384–1394. © The Author(s) 2016

Blood Cancer Journal

Você também pode gostar